Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mult Scler ; 28(8): 1179-1188, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34841955

RESUMEN

BACKGROUND: Cell-based therapies for multiple sclerosis (MS), including those employing autologous bone marrow-derived mesenchymal stromal cells (MSC) are being examined in clinical trials. However, recent studies have identified abnormalities in the MS bone marrow microenvironment. OBJECTIVE: We aimed to compare the secretome of MSC isolated from control subjects (C-MSC) and people with MS (MS-MSC) and explore the functional relevance of findings. METHODS: We employed high throughput proteomic analysis, enzyme-linked immunosorbent assays and immunoblotting, as well as in vitro assays of enzyme activity and neuroprotection. RESULTS: We demonstrated that, in progressive MS, the MSC secretome has lower levels of mitochondrial fumarate hydratase (mFH). Exogenous mFH restores the in vitro neuroprotective potential of MS-MSC. Furthermore, MS-MSC expresses reduced levels of fumarate hydratase (FH) with downstream reduction in expression of master regulators of oxidative stress. CONCLUSIONS: Our findings are further evidence of dysregulation of the bone marrow microenvironment in progressive MS with respect to anti-oxidative capacity and immunoregulatory potential. Given the clinical utility of the fumaric acid ester dimethyl fumarate in relapsing-remitting MS, our findings have potential implication for understanding MS pathophysiology and personalised therapeutic intervention.


Asunto(s)
Fumarato Hidratasa , Células Madre Mesenquimatosas , Mitocondrias , Esclerosis Múltiple Crónica Progresiva , Neuroprotección , Fumarato Hidratasa/metabolismo , Humanos , Mitocondrias/enzimología , Esclerosis Múltiple Crónica Progresiva/metabolismo , Proteómica
2.
J Pathol ; 247(4): 422-434, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30565681

RESUMEN

The overall survival for patients with primary glioblastoma is very poor. Glioblastoma contains a subpopulation of glioma stem cells (GSC) that are responsible for tumour initiation, treatment resistance and recurrence. PPARα is a transcription factor involved in the control of lipid, carbohydrate and amino acid metabolism. We have recently shown that PPARα gene and protein expression is increased in glioblastoma and has independent clinical prognostic significance in multivariate analyses. In this work, we report that PPARα is overexpressed in GSC compared to foetal neural stem cells. To investigate the role of PPARα in GSC, we knocked down its expression using lentiviral transduction with short hairpin RNA (shRNA). Transduced GSC were tagged with luciferase and stereotactically xenografted into the striatum of NOD-SCID mice. Bioluminescent and magnetic resonance imaging showed that knockdown (KD) of PPARα reduced the tumourigenicity of GSC in vivo. PPARα-expressing control GSC xenografts formed invasive histological phenocopies of human glioblastoma, whereas PPARα KD GSC xenografts failed to establish viable intracranial tumours. PPARα KD GSC showed significantly reduced proliferative capacity and clonogenic potential in vitro with an increase in cellular senescence. In addition, PPARα KD resulted in significant downregulation of the stem cell factors c-Myc, nestin and SOX2. This was accompanied by downregulation of the PPARα-target genes and key regulators of fatty acid oxygenation ACOX1 and CPT1A, with no compensatory increase in glycolytic flux. These data establish the aberrant overexpression of PPARα in GSC and demonstrate that this expression functions as an important regulator of tumourigenesis, linking self-renewal and the malignant phenotype in this aggressive cancer stem cell subpopulation. We conclude that targeting GSC PPARα expression may be a therapeutically beneficial strategy with translational potential as an adjuvant treatment. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , PPAR alfa/metabolismo , ARN Interferente Pequeño/farmacología , Animales , Biomarcadores de Tumor/metabolismo , Transformación Celular Neoplásica , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen/métodos , Humanos , Lentivirus , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/patología , Fenotipo , Transducción de Señal/fisiología , Trasplante Heterólogo , Células Tumorales Cultivadas
3.
Ann Neurol ; 83(4): 779-793, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29534309

RESUMEN

OBJECTIVE: Friedreich's ataxia is an incurable inherited neurological disease caused by frataxin deficiency. Here, we report the neuroreparative effects of myeloablative allogeneic bone marrow transplantation in a humanized murine model of the disease. METHODS: Mice received a transplant of fluorescently tagged sex-mismatched bone marrow cells expressing wild-type frataxin and were assessed at monthly intervals using a range of behavioral motor performance tests. At 6 months post-transplant, mice were euthanized for protein and histological analysis. In an attempt to augment numbers of bone marrow-derived cells integrating within the nervous system and improve therapeutic efficacy, a subgroup of transplanted mice also received monthly subcutaneous infusions of the cytokines granulocyte-colony stimulating factor and stem cell factor. RESULTS: Transplantation caused improvements in several indicators of motor coordination and locomotor activity. Elevations in frataxin levels and antioxidant defenses were detected. Abrogation of disease pathology throughout the nervous system was apparent, together with extensive integration of bone marrow-derived cells in areas of nervous tissue injury that contributed genetic material to mature neurons, satellite-like cells, and myelinating Schwann cells by processes including cell fusion. Elevations in circulating bone marrow-derived cell numbers were detected after cytokine administration and were associated with increased frequencies of Purkinje cell fusion and bone marrow-derived dorsal root ganglion satellite-like cells. Further improvements in motor coordination and activity were evident. INTERPRETATION: Our data provide proof of concept of gene replacement therapy, via allogeneic bone marrow transplantation, that reverses neurological features of Friedreich's ataxia with the potential for rapid clinical translation. Ann Neurol 2018;83:779-793.


Asunto(s)
Trasplante de Médula Ósea/métodos , Ataxia de Friedreich/cirugía , Animales , Peso Corporal/fisiología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Conducta Exploratoria/fisiología , Ataxia de Friedreich/genética , Ganglios Espinales/patología , Factor Estimulante de Colonias de Granulocitos/metabolismo , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Unión a Hierro/genética , Leucocitos Mononucleares/patología , Ratones , Ratones Endogámicos C57BL , Fuerza Muscular/fisiología , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/patología , Frataxina
4.
Ann Neurol ; 81(2): 212-226, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28009062

RESUMEN

OBJECTIVES: Friedreich's ataxia is a devastating neurological disease currently lacking any proven treatment. We studied the neuroprotective effects of the cytokines, granulocyte-colony stimulating factor (G-CSF) and stem cell factor (SCF) in a humanized murine model of Friedreich's ataxia. METHODS: Mice received monthly subcutaneous infusions of cytokines while also being assessed at monthly time points using an extensive range of behavioral motor performance tests. After 6 months of treatment, neurophysiological evaluation of both sensory and motor nerve conduction was performed. Subsequently, mice were sacrificed for messenger RNA, protein, and histological analysis of the dorsal root ganglia, spinal cord, and cerebellum. RESULTS: Cytokine administration resulted in significant reversal of biochemical, neuropathological, neurophysiological, and behavioural deficits associated with Friedreich's ataxia. Both G-CSF and SCF had pronounced effects on frataxin levels (the primary molecular defect in the pathogenesis of the disease) and a regulators of frataxin expression. Sustained improvements in motor coordination and locomotor activity were observed, even after onset of neurological symptoms. Treatment also restored the duration of sensory nerve compound potentials. Improvements in peripheral nerve conduction positively correlated with cytokine-induced increases in frataxin expression, providing a link between increases in frataxin and neurophysiological function. Abrogation of disease-related pathology was also evident, with reductions in inflammation/gliosis and increased neural stem cell numbers in areas of tissue injury. INTERPRETATION: These experiments show that cytokines already clinically used in other conditions offer the prospect of a novel, rapidly translatable, disease-modifying, and neuroprotective treatment for Friedreich's ataxia. Ann Neurol 2017;81:212-226.


Asunto(s)
Conducta Animal/efectos de los fármacos , Ataxia de Friedreich/tratamiento farmacológico , Factor Estimulante de Colonias de Granulocitos/farmacología , Proteínas de Unión a Hierro/metabolismo , Conducción Nerviosa/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Nervios Periféricos/efectos de los fármacos , Factor de Células Madre/farmacología , Animales , Modelos Animales de Enfermedad , Ataxia de Friedreich/metabolismo , Ataxia de Friedreich/fisiopatología , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fármacos Neuroprotectores/administración & dosificación , Factor de Células Madre/administración & dosificación , Frataxina
5.
Cytotherapy ; 20(1): 21-28, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28917625

RESUMEN

BACKGROUND: Clinical trials using ex vivo expansion of autologous mesenchymal stromal cells (MSCs) are in progress for several neurological diseases including multiple sclerosis (MS). Given that environment alters MSC function, we examined whether in vitro expansion, increasing donor age and progressive MS affect the neuroprotective properties of the MSC secretome. METHODS: Comparative analyses of neuronal survival in the presence of MSC-conditioned medium (MSCcm) isolated from control subjects (C-MSCcm) and those with MS (MS-MSCcm) were performed following (1) trophic factor withdrawal and (2) nitric oxide-induced neurotoxicity. RESULTS: Reduced neuronal survival following trophic factor withdrawal was seen in association with increasing expansion of MSCs in vitro and MSC donor age. Controlling for these factors, there was an independent, negative effect of progressive MS. In nitric oxide neurotoxicity, MSCcm-mediated neuroprotection was reduced when C-MSCcm was isolated from higher-passage MSCs and was negatively associated with increasing MSC passage number and donor age. Furthermore, the neuroprotective effect of MSCcm was lost when MSCs were isolated from patients with MS. DISCUSSION: Our findings have significant implications for MSC-based therapy in neurodegenerative conditions, particularly for autologous MSC therapy in MS. Impaired neuroprotection mediated by the MSC secretome in progressive MS may reflect reduced reparative potential of autologous MSC-based therapy in MS and it is likely that the causes must be addressed before the full potential of MSC-based therapy is realized. Additionally, we anticipate that understanding the mechanisms responsible will contribute new insights into MS pathogenesis and may also be of wider relevance to other neurodegenerative conditions.


Asunto(s)
Envejecimiento/patología , Progresión de la Enfermedad , Células Madre Mesenquimatosas/metabolismo , Esclerosis Múltiple/patología , Esclerosis Múltiple/terapia , Fármacos Neuroprotectores/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Humanos , Persona de Mediana Edad , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Neuroprotección/efectos de los fármacos , Óxido Nítrico/metabolismo
6.
Mult Scler ; 24(7): 919-931, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-28548004

RESUMEN

BACKGROUND: Autologous bone-marrow-derived cells are currently employed in clinical studies of cell-based therapy in multiple sclerosis (MS) although the bone marrow microenvironment and marrow-derived cells isolated from patients with MS have not been extensively characterised. OBJECTIVES: To examine the bone marrow microenvironment and assess the proliferative potential of multipotent mesenchymal stromal cells (MSCs) in progressive MS. METHODS: Comparative phenotypic analysis of bone marrow and marrow-derived MSCs isolated from patients with progressive MS and control subjects was undertaken. RESULTS: In MS marrow, there was an interstitial infiltrate of inflammatory cells with lymphoid (predominantly T-cell) nodules although total cellularity was reduced. Controlling for age, MSCs isolated from patients with MS had reduced in vitro expansion potential as determined by population doubling time, colony-forming unit assay, and expression of ß-galactosidase. MS MSCs expressed reduced levels of Stro-1 and displayed accelerated shortening of telomere terminal restriction fragments (TRF) in vitro. CONCLUSION: Our results are consistent with reduced proliferative capacity and ex vivo premature ageing of bone-marrow-derived cells, particularly MSCs, in MS. They have significant implication for MSC-based therapies for MS and suggest that accelerated cellular ageing and senescence may contribute to the pathophysiology of progressive MS.


Asunto(s)
Proliferación Celular , Senescencia Celular , Células Madre Mesenquimatosas/patología , Esclerosis Múltiple/patología , Adulto , Proliferación Celular/fisiología , Células Cultivadas , Senescencia Celular/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Nicho de Células Madre/fisiología
7.
Histopathology ; 70(7): 1030-1043, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27926792

RESUMEN

AIMS: PPARα agonists are in current clinical use as hypolipidaemic agents and show significant antineoplastic effects in human glioblastoma models. To date however, the expression of PPARα in large-scale glioblastoma datasets has not been examined. We aimed to investigate the expression of the transcription factor PPARα in primary glioblastoma, the relationship between PPARα expression and patients' clinicopathological features and other molecular markers associated with gliomagenesis. METHODS AND RESULTS: With protein immunoblotting techniques and reverse transcription quantitative real-time PCR, PPARα was found to be significantly overexpressed in glioblastoma compared with control brain tissue (P = 0.032 and P = 0.005). PPARA gene expression was found to be enriched in the classical glioblastoma subtype within The Cancer Genome Atlas (TCGA) dataset. Although not associated with overall survival when assessed by immunohistochemistry, cross-validation with the TCGA dataset and multivariate analyses identified PPARA gene expression as an independent prognostic marker for overall survival (P = 0.042). Finally, hierarchical clustering revealed novel, significant associations between high PPARA expression and a putative set of glioblastoma molecular mediators including EMX2, AQP4, and NTRK2. CONCLUSIONS: PPARα is overexpressed in primary glioblastoma and high PPARA expression functions as an independent prognostic marker in the glioblastoma TCGA dataset. Further studies are required to explore genetic associations with high PPARA expression and to analyse the predictive role of PPARα expression in glioblastoma models in response to PPARα agonists.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias Encefálicas/patología , Glioblastoma/patología , PPAR alfa/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidad , Niño , Femenino , Glioblastoma/metabolismo , Glioblastoma/mortalidad , Humanos , Isocitrato Deshidrogenasa/genética , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , PPAR alfa/análisis , Pronóstico , Modelos de Riesgos Proporcionales
8.
J Neurosci Res ; 93(6): 882-92, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25639260

RESUMEN

Axonal injury is often characterized by axonal transport defects and abnormal accumulation of intra-axonal components. Nitric oxide (NO) has a key role in mediating inflammatory axonopathy in many neurodegenerative diseases, but little is known about how nitrosative/oxidative stress affects axonal transport or whether reductions in kinesin superfamily protein (KIF) expression correlate with axon pathology. KIFs are molecular motors that have a key role in axonal and dendritic transport, and impairment of these mechanisms has been associated with a number of neurological disorders. This study shows that rat cortical neurons exposed to NO display both a time-dependent decrease in KIF gene/protein expression and neurofilament phosphorylation in addition to a reduction in axonal length and neuronal survival. Because mesenchymal stem cells (MSCs) represent a promising therapeutic candidate for neuronal/axonal repair, this study analyzes the capacity of MSCs to protect neurons and axonal transport mechanisms from NO damage. Results show that coculture of MSCs with NO-exposed neurons results in the preservation of KIF expression, axonal length, and neuronal survival. Altogether, these results suggest a potential mechanism involved in the disruption of axonal transport and abnormal accumulation of proteins in axons during nitrosative insult. We hypothesize that impaired axonal transport contributes, per se, to progression of injury and provide further evidence of the therapeutic potential of MSCs for neurodegenerative disorders.


Asunto(s)
Axones/patología , Depuradores de Radicales Libres/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Cinesinas/metabolismo , Neuronas/efectos de los fármacos , Óxido Nítrico/toxicidad , Análisis de Varianza , Animales , Antígenos CD/metabolismo , Axones/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Técnicas de Cocultivo , Embrión de Mamíferos , Humanos , Cinesinas/genética , Proteínas de Neurofilamentos/metabolismo , Ratas , Factores de Tiempo
9.
Public Health Nutr ; 18(2): 275-85, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24698168

RESUMEN

OBJECTIVE: To assess the agreement between self-reported and parent-reported dietary and physical activity habits in children; and to evaluate the socio-economic determinants of healthier habits (Mediterranean diet and physical activity) among children. DESIGN: Cross-sectional analysis of children recruited to a cluster-randomized controlled trial (Program SI!). Information about children's and parents' dietary and physical activity habits was obtained through validated questionnaires (Program SI! questionnaires, Kidmed, Krece Plus and Predimed scores). SETTING: Twenty-four schools in Madrid, Spain. SUBJECTS: Children (n 2062) aged 3-5 years and their parents (n 1949). RESULTS: There was positive agreement between parental- and self-reporting for three of the six children's habits examined. Parents' dietary and physical activity patterns were associated with those of their children. The main determinants of higher scores in children were higher parental age, the mother's scores, Spanish origin and higher awareness of human health (P<0·005). Children from parents with a low educational level had lower odds for scoring positively on items such as using olive oil (OR=0·23; 95 % CI 0·13, 0·41) and not skipping breakfast (OR=0·36; 95 % CI 0·23, 0·55), but higher odds for meeting the recommendations for consuming pulses (OR=1·71; 95 % CI 1·14, 2·55). Other habits being influenced by parental socio-economic status included the consumption of vegetables, fish, nuts, avoidance of fast food, and consumption of bakery products for breakfast. CONCLUSIONS: Children's habits may be influenced by their parents' health awareness and other socio-economic characteristics. These findings suggest that intervention strategies, even in very young children, should also target parents in order to achieve maximum success.


Asunto(s)
Conducta Infantil , Fenómenos Fisiológicos Nutricionales Infantiles , Dieta , Conducta Alimentaria , Hábitos , Actividad Motora , Preescolar , Estudios Transversales , Dieta Mediterránea , Femenino , Humanos , Masculino , Conducta Materna , Política Nutricional , Padres , Conducta Paterna , Cooperación del Paciente , Instituciones Académicas , Autoinforme , Factores Socioeconómicos , España , Población Urbana
10.
Mult Scler ; 20(6): 651-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24067896

RESUMEN

BACKGROUND: Peroxisomes are organelles in eukaryotic cells with multiple functions including the detoxification of reactive oxygen species, plasmalogen synthesis and ß-oxidation of fatty acids. Recent evidence has implicated peroxisomal dysfunction in models of multiple sclerosis (MS) disease progression. OBJECTIVES: Our aims were to determine whether there are changes in peroxisomes in MS grey matter (GM) compared to control GM. METHODS: We analysed cases of MS and control GM immunocytochemically to assess peroxisomal membrane protein (PMP70) and neuronal proteins. We examined the expression of ABCD3 (the gene that encodes PMP70) in MS and control GM. Analyses of very long chain fatty acid (VLCFA) levels in GM were performed. RESULTS: PMP70 immunolabelling of neuronal somata was significantly lower in MS GM compared to control. Calibration of ABCD3 gene expression with reference to glyceraldehyde 3-phsophate dehydrogenase (GAPDH) revealed overall decreases in expression in MS compared to controls. Mean PMP70 counts in involved MS GM negatively correlated to disease duration. Elevations in C26:0 (hexacosanoic acid) were found in MS GM. CONCLUSIONS: Collectively, these observations provide evidence that there is an overall reduction in peroxisomal gene expression and peroxisomal proteins in GM neurons in MS. Changes in peroxisomal function may contribute to neuronal dysfunction and degeneration in MS.


Asunto(s)
Sustancia Gris/patología , Proteínas de la Membrana/metabolismo , Esclerosis Múltiple/patología , Neuronas/patología , Peroxisomas/patología , Transportadoras de Casetes de Unión a ATP/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Expresión Génica/fisiología , Sustancia Gris/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Neuronas/metabolismo
11.
Stem Cells Transl Med ; 13(3): 219-229, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38097199

RESUMEN

Bone marrow (BM)-derived mesenchymal stromal cells (MSCs) are promising candidates for cell-based therapy for several immune-mediated inflammatory diseases (IMIDs) due to their multiplicity of immunomodulatory and reparative properties and favorable safety profile. However, although preclinical data were encouraging, the clinical benefit demonstrated in clinical trials of autologous MSC transplantation in a number of conditions has been less robust. This may be explained by the growing body of evidence pointing to abnormalities of the bone marrow microenvironment in IMIDs, including impaired MSC function. However, it is not currently known whether these abnormalities arise as a cause or consequence of disease, the role they play in disease initiation and/or progression, or whether they themselves are targets for disease modification. Here, we review current knowledge about the function of the BM microenvironment in IMIDs including multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, and type I diabetes, focusing on MSCs in particular. We predict that an improved understanding of disease-related changes in the bone marrow microenvironment including the role of MSCs in vivo, will yield new insights into pathophysiology and aid identification of new drug targets and optimization of cell-based therapy in IMIDs.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Médula Ósea , Células de la Médula Ósea , Agentes Inmunomoduladores , Células Madre Mesenquimatosas/fisiología , Trasplante Autólogo , Humanos
12.
Cell Mol Biol Lett ; 18(2): 163-86, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23430457

RESUMEN

Mesenchymal stem cells (MSCs) are multipotent cells that are able to differentiate into mesodermal lineages (osteogenic, adipogenic, chondrogenic), but also towards non-mesodermal derivatives (e.g. neural cells). Recent in vitro studies revealed that, in the absence of any kind of differentiation stimuli, undifferentiated MSCs express neural differentiation markers, but the literature data do not all concur. Considering their promising therapeutic potential for neurodegenerative diseases, it is very important to expand our knowledge about this particular biological property of MSCs. In this study, we confirmed the spontaneous expression of neural markers (neuronal, glial and progenitor markers) by undifferentiated human MSCs (hMSCs) and in particular, we demonstrated that the neuronal markers ßIII-tubulin and NeuN are expressed by a very high percentage of hMSCs, regardless of the number of culture passages and the culture conditions. Moreover, the neuronal markers ßIII-tubulin and NeuN are still expressed by hMSCs after in vitro osteogenic and adipogenic differentiation. On the other hand, chondrogenically differentiated hMSCs are negative for these markers. Our findings suggest that the expression of neuronal markers could be common to a wide range of cellular types and not exclusive for neuronal lineages. Therefore, the expression of neuronal markers alone is not sufficient to demonstrate the differentiation of MSCs towards the neuronal phenotype. Functional properties analysis is also required.


Asunto(s)
Adipogénesis , Biomarcadores/metabolismo , Diferenciación Celular , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Neuronas/metabolismo , Osteogénesis , Linaje de la Célula , Separación Celular , Células Cultivadas , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Humanos , Immunoblotting , Mesodermo/metabolismo , Neuraminidasa/metabolismo , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Tubulina (Proteína)/metabolismo
13.
BMC Public Health ; 13: 1208, 2013 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-24359285

RESUMEN

BACKGROUND: Unhealthy lifestyles contribute to the development of cardiovascular risk factors, whose incidence is increasing among children and adolescents. The Program SI! is a long-term, multi-target behavioral intervention to promote healthy lifestyle habits in children through the school environment. The objective of the study is to evaluate the efficacy of this intervention in its first phase, preschoolers. METHODS: Cluster-randomized controlled trial in public schools in the city of Madrid, Spain. A total 24 schools, including 2062 children (3-5 years), 1949 families, and 125 teachers participated in the study. Schools were assigned to their usual school curriculum or to engage in an additional multi-component intervention (Program SI!). The primary outcome of this trial is 1-school year changes from baseline in scores for children's knowledge, attitudes and habits (KAH). Secondary outcomes are 1-school year changes from baseline in scores for knowledge, attitudes, and habits among parents, teachers, and the school environment. RESULTS: After 1-school year, our results indicate that the Program SI! intervention increases children's KAH scores, both overall (3.45, 95% CI, 1.84-5.05) and component-specific (Diet: 0.93, 95% CI, 0.12-1.75; Physical activity: 1.93, 95% CI, 1.17-2.69; Human body: 0.65, 95% CI, 0.07-1.24) score. CONCLUSIONS: The Program SI! is demonstrated as an effective and feasible strategy for increasing knowledge and improving lifestyle attitudes and habits among very young children. TRIAL REGISTRATION: NCT01579708, Evaluation of the Program SI! for Preschool Education: A School-Based Randomized Controlled Trial (Preschool-SI!).


Asunto(s)
Conducta Infantil/psicología , Promoción de la Salud/métodos , Estilo de Vida , Servicios de Salud Escolar/organización & administración , Preescolar , Análisis por Conglomerados , Estudios de Factibilidad , Conductas Relacionadas con la Salud , Conocimientos, Actitudes y Práctica en Salud , Humanos , Evaluación de Programas y Proyectos de Salud , Medio Social , España
14.
BMC Public Health ; 13: 656, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23855415

RESUMEN

BACKGROUND: The onset of inadequate behaviors leading to the development of risk factors for chronic diseases is known to occur early in life. An effective program for health promotion should therefore focus on children and their environment, as the starting point for behavior development. The overarching objective of the Program SI! (Salud Integral - Comprehensive Health) is to intervene at the school level, to establish and develop life-lasting habits that will help preserving health during adulthood. The Program SI! comprises five consecutive subprograms according to the five stages of education in Spain, the first being in preschoolers. This study aims to evaluate the efficacy of Program SI! to establish and improve lifestyle behaviors in children (preschoolers aged 3-5 years), their parents, and teachers, and also improving the school environment. A secondary objective is to evaluate improvements in cardiovascular health-related markers (anthropometric parameters, blood pressure, and dietary and physical activity patterns) in these same children. METHODS/DESIGN: 24 public schools from the city of Madrid (Spain) were allocated through stratified randomization to intervention or control. The intervention schools follow the Program SI!, which provides didactic units, emotions cards, healthy tips, and online resources. The intervention schools integrate the Program SI! into their scholar curriculum organized in four complete weeks during each academic year during the 3 years of preschool education. Control schools follow their normal curriculum. Primary outcomes are 1-year, and 3-year changes from baseline of scores for knowledge, attitudes, and habits (KAH) of children, their parents and teachers in regards to a healthy lifestyle. Secondary outcomes are 1-year, and 3-year changes from baseline in clinical and anthropometric parameters of children. DISCUSSION: The Program SI! is a long-term health promotion program starting in 3 years old. It incorporates the traditional areas of intervention (diet and physical activity), introducing additional components such as knowledge of the human body and management of emotions to achieve a comprehensive intervention. The Program SI! is designed to be an effective, sustainable health promotion program for the adoption of healthy behaviors from early in life. TRIAL REGISTRATION NUMBER: NCT01579708.


Asunto(s)
Curriculum , Conductas Relacionadas con la Salud , Promoción de la Salud/métodos , Obesidad/prevención & control , Evaluación de Programas y Proyectos de Salud , Servicios de Salud Escolar , Instituciones Académicas , Adulto , Antropometría , Terapia Conductista , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/prevención & control , Preescolar , Dieta , Emociones , Ejercicio Físico , Conducta Alimentaria , Femenino , Conocimientos, Actitudes y Práctica en Salud , Humanos , Estilo de Vida , Masculino , Obesidad/etiología , Padres/educación , Factores de Riesgo , España
15.
J Biomed Biotechnol ; 2012: 820821, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23093867

RESUMEN

The spontaneous expression of neural markers by mesenchymal stem cells (MSCs) has been considered to be a demonstration of MSCs' predisposition to differentiate towards neural lineages. In view of their application in cell therapy for neurodegenerative diseases, it is very important to deepen the knowledge about this distinctive biological property of MSCs. In this study, we evaluated the expression of neuronal and glial markers in undifferentiated rat MSCs (rMSCs) at different culture passages (from early to late). rMSCs spontaneously expressed neural markers depending on culture passage, and they were coexpressed or not with the neural progenitor marker nestin. In contrast, the number of rMSCs expressing mesengenic differentiation markers was very low or even completely absent. Moreover, rMSCs at late culture passages were not senescent cells and maintained the MSC immunophenotype. However, their differentiation capabilities were altered. In conclusion, our results support the concept of MSCs as multidifferentiated cells and suggest the existence of immature and mature neurally fated rMSC subpopulations. A possible correlation between specific MSC subpopulations and specific neural lineages could optimize the use of MSCs in cell transplantation therapy for the treatment of neurological diseases.


Asunto(s)
Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/fisiología , Neuronas/citología , Neuronas/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Ratas , Ratas Sprague-Dawley
16.
Nat Commun ; 13(1): 4655, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35945193

RESUMEN

Friedreich's ataxia (FA) is an inherited progressive neurodegenerative disease for which there is no proven disease-modifying treatment. Here we perform an open-label, pilot study of recombinant human granulocyte-colony stimulating factor (G-CSF) administration in seven people with FA (EudraCT: 2017-003084-34); each participant receiving a single course of G-CSF (Lenograstim; 1.28 million units per kg per day for 5 days). The primary outcome is peripheral blood mononuclear cell frataxin levels over a 19-day period. The secondary outcomes include safety, haematopoietic stem cell (HSC) mobilisation, antioxidant levels and mitochondrial enzyme activity. The trial meets pre-specified endpoints. We show that administration of G-CSF to people with FA is safe. Mobilisation of HSCs in response to G-CSF is comparable to that of healthy individuals. Notably, sustained increases in cellular frataxin concentrations and raised PGC-1α and Nrf2 expression are detected. Our findings show potential for G-CSF therapy to have a clinical impact in people with FA.


Asunto(s)
Ataxia de Friedreich , Factor Estimulante de Colonias de Granulocitos , Proteínas Recombinantes , Ataxia de Friedreich/tratamiento farmacológico , Factor Estimulante de Colonias de Granulocitos/efectos adversos , Granulocitos/metabolismo , Humanos , Leucocitos Mononucleares/metabolismo , Proyectos Piloto , Proteínas Recombinantes/efectos adversos
17.
Stem Cells Transl Med ; 7(10): 748-758, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30063300

RESUMEN

The potential of autologous cell-based therapies including those using multipotent mesenchymal stromal cells (MSCs) is being investigated for multiple sclerosis (MS) and other neurological conditions. However, the phenotype of MSC in neurological diseases has not been fully characterized. We have previously shown that MSC isolated from patients with progressive MS (MS-MSC) have reduced expansion potential, premature senescence, and reduced neuroprotective potential in vitro. In view of the role of antioxidants in ageing and neuroprotection, we examined the antioxidant capacity of MS-MSC demonstrating that MS-MSC secretion of antioxidants superoxide dismutase 1 (SOD1) and glutathione S-transferase P (GSTP) is reduced and correlates negatively with the duration of progressive phase of MS. We confirmed reduced expression of SOD1 and GSTP by MS-MSC along with reduced activity of SOD and GST and, to examine the antioxidant capacity of MS-MSC under conditions of nitrosative stress, we established an in vitro cell survival assay using nitric oxide-induced cell death. MS-MSC displayed differential susceptibility to nitrosative stress with accelerated senescence and greater decline in expression of SOD1 and GSTP in keeping with reduced expression of master regulators of antioxidant responses nuclear factor erythroid 2-related factor 2 and peroxisome proliferator-activated receptor gamma coactivator 1-α. Our results are compatible with dysregulation of antioxidant responses in MS-MSC and have significant implications for development of autologous MSC-based therapies for MS, optimization of which may require that these functional deficits are reversed. Furthermore, improved understanding of the underlying mechanisms may yield novel insights into MS pathophysiology and biomarker identification. Stem Cells Translational Medicine 2018;7:748-758.


Asunto(s)
Antioxidantes/metabolismo , Células Madre Mesenquimatosas/metabolismo , Esclerosis Múltiple/patología , Células de la Médula Ósea/citología , Senescencia Celular/efectos de los fármacos , Femenino , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Humanos , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Persona de Mediana Edad , Esclerosis Múltiple/terapia , Factor 2 Relacionado con NF-E2/metabolismo , Compuestos Nitrosos/farmacología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo
18.
J Clin Pathol ; 71(8): 695-701, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29463577

RESUMEN

AIMS: Histopathological tissue samples are being increasingly used as sources of nucleic acids in molecular pathology translational research. This study investigated the suitability of glioblastoma and control central nervous system (CNS) formalin-fixed paraffin embedded (FFPE) tissue-derived RNA for gene expression analyses. METHODS: Total RNA was extracted from control (temporal lobe resection tissue) and glioblastoma FFPE tissue samples. RNA purity (260/280 ratios) was determined and RNA integrity number (RIN) analysis was performed. RNA was subsequently used for RT-qPCR for two reference genes, 18S and GAPDH. RESULTS: Reference gene expression was equivalent between control and glioblastoma tissue when using RNA extracted from FFPE tissue, which has key implications for biological normalisation for CNS gene expression studies. There was a significant difference between the mean RIN values of control and glioblastoma FFPE tissue. There was no significant correlation between 260/280 or RIN values versus total RNA yield. The age of the tissue blocks did not influence RNA yield, fragmentation or purity. There was no significant correlation between RIN or 260/280 ratios and mean qPCR cycle threshold for either reference gene. CONCLUSIONS: This study showed that routinely available CNS FFPE tissue is suitable for RNA extraction and downstream gene expression studies, even after 60 months of storage. Substantial RNA fragmentation associated with glioblastoma and control FFPE tissue blocks did not preclude downstream RT-qPCR gene expression analyses. Cross validation with both archival and prospectively collated FFPE specimens is required to further demonstrate that CNS tissue blocks can be used in novel translational molecular biomarker studies.


Asunto(s)
Neoplasias Encefálicas/genética , Epilepsia del Lóbulo Temporal/genética , Fijadores/química , Formaldehído/química , Perfilación de la Expresión Génica , Glioblastoma/genética , Adhesión en Parafina , Estabilidad del ARN , ARN Neoplásico/genética , Fijación del Tejido/métodos , Neoplasias Encefálicas/cirugía , Estudios de Casos y Controles , Epilepsia del Lóbulo Temporal/cirugía , Perfilación de la Expresión Génica/normas , Glioblastoma/cirugía , Humanos , Adhesión en Parafina/normas , Valor Predictivo de las Pruebas , Control de Calidad , Reproducibilidad de los Resultados , Factores de Tiempo , Fijación del Tejido/normas
19.
Eur J Pharmacol ; 538(1-3): 115-23, 2006 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-16631733

RESUMEN

The aim was to determine whether high glucose levels interfere with nitric oxide (NO) production and inducible NO synthase (iNOS) protein expression in interleukin-1beta-stimulated vascular smooth muscle cells from normotensive Wistar Kyoto and spontaneously hypertensive rats. Cells were incubated with either normal (5.5 mM) or high (22 mM) d-glucose for 72 h and with interleukin-1beta (10 ng/ml) for the last 24 h. High glucose increased nitrite levels, iNOS expression and protein kinase C activity in cells from normotensive rats and had no effect in cells from hypertensive rats. High glucose effects on nitrite production and iNOS expression was abolished by the selective inhibitor for the protein kinase C-betaII, 5,21:12,17-dimetheno-18H-dibenzo[i,o]pyrrolo[3,4-1] [1,8]diacyclohexadecine-18,20 (19H)-dione, 8-[(dimethylamino) methyl]-6,7,8,9,10,11-hexahydro-monomethanesulfonate (LY379196, 30 nM). Calphostin C (1 microM) and LY379196 (10 microM) reduced nitrite levels and iNOS expression only in cells from normotensive rats treated with both media. These results suggest that high glucose increases inducible nitric oxide synthase induction and subsequent NO production by activating the protein kinase C-betaII; this mechanism seems to be altered in hypertension.


Asunto(s)
Glucosa/farmacología , Interleucina-1/farmacología , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Animales , Aorta Torácica/citología , Western Blotting , Células Cultivadas , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Masculino , Mesilatos/farmacología , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Naftalenos/farmacología , Óxido Nítrico/biosíntesis , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Proteína Quinasa C/fisiología , Proteína Quinasa C beta , Pirroles/farmacología , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Acetato de Tetradecanoilforbol/farmacología
20.
Brain Res ; 1642: 452-460, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27086975

RESUMEN

Cerebellar dysfunction is a significant contributor to disability in multiple sclerosis (MS). Both white matter (WM) and grey matter (GM) injury occurs within MS cerebellum and, within GM, demyelination, inflammatory cell infiltration and neuronal injury contribute to on-going pathology. The precise nature of cerebellar GM injury is, however, unknown. Oxidative stress pathways with ultimate lipid peroxidation and cell membrane injury occur extensively in MS and the purpose of this study was to investigate these processes in MS cerebellar GM. Post-mortem human cerebellar GM from MS and control subjects was analysed immunohistochemically, followed by semi-quantitative analysis of markers of cellular injury, lipid peroxidation and anti-oxidant enzyme expression. We have shown evidence for reduction in myelin and neuronal markers in MS GM, coupled to an increase in expression of a microglial marker. We also show that the lipid peroxidation product 4-hydroxynonenal co-localises with myelin and its levels negatively correlate to myelin basic protein levels. Furthermore, superoxide dismutase (SOD1 and 2) enzymes, localised within cerebellar neurons, are up-regulated, yet the activation of subsequent enzymes responsible for the detoxification of hydrogen peroxide, catalase and glutathione peroxidase are relatively deficient. These studies provide evidence for oxidative injury in MS cerebellar GM and further help define disease mechanisms within the MS brain.


Asunto(s)
Cerebelo/metabolismo , Cerebelo/patología , Sustancia Gris/metabolismo , Sustancia Gris/patología , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Adulto , Anciano , Anciano de 80 o más Años , Aldehídos/metabolismo , Femenino , Humanos , Peroxidación de Lípido/fisiología , Masculino , Microglía/metabolismo , Microglía/patología , Persona de Mediana Edad , Proteína Básica de Mielina/metabolismo , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Neuronas/metabolismo , Neuronas/patología , Estrés Oxidativo/fisiología , ARN Mensajero/metabolismo , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1/metabolismo , Sustancia Blanca/metabolismo , Sustancia Blanca/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA